AU2012324824B2 - Processes for the preparation of 6-Chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and of its precursors - Google Patents

Processes for the preparation of 6-Chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and of its precursors Download PDF

Info

Publication number
AU2012324824B2
AU2012324824B2 AU2012324824A AU2012324824A AU2012324824B2 AU 2012324824 B2 AU2012324824 B2 AU 2012324824B2 AU 2012324824 A AU2012324824 A AU 2012324824A AU 2012324824 A AU2012324824 A AU 2012324824A AU 2012324824 B2 AU2012324824 B2 AU 2012324824B2
Authority
AU
Australia
Prior art keywords
compound
process according
solvent
reaction
water
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
AU2012324824A
Other versions
AU2012324824A1 (en
Inventor
Enrica DIODATO
Katia Marcucci
Martin Rettig
Huw Roberts
Russell Thomas
Paul WIEDENAU
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AOP Orphan IP AG
Original Assignee
Ipsol AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ipsol AG filed Critical Ipsol AG
Publication of AU2012324824A1 publication Critical patent/AU2012324824A1/en
Application granted granted Critical
Publication of AU2012324824B2 publication Critical patent/AU2012324824B2/en
Assigned to AOP Orphan Pharmaceuticals reassignment AOP Orphan Pharmaceuticals Request for Assignment Assignors: SIENA BIOTECH S.P.A.
Assigned to IPSOL AG reassignment IPSOL AG Request for Assignment Assignors: AOP Orphan Pharmaceuticals
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/56Ring systems containing three or more rings
    • C07D209/80[b, c]- or [b, d]-condensed
    • C07D209/82Carbazoles; Hydrogenated carbazoles
    • C07D209/88Carbazoles; Hydrogenated carbazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the ring system

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Indole Compounds (AREA)

Abstract

The invention relates to a novel process for the preparation of rac-6-Chloro-2,3,4,9-tetrahydro-1

Description

PROCESSES FOR THE PREPARATION OF 6 - CHLORO -2,3,4,9- TETRAHYDRO - 1H -CARBAZOLE -1 - CARBOXAMIDE AND OF ITS PRECURSORS
The present invention relates to a novel process for the preparation of rac-6-Chloro-2,3,4,9-tetrahydro- 17/-carbazole-1 -carboxamide (I) in pharmaceutically acceptable polymorphic form and to intermediates thereof.
BACKGROUND TO THE INVENTION
The compound 6-Chloro-2,3,4,9-tetrahydro-lH-carbazole-l-carboxamide (I) is known from W02005026112 to possess anti Sirtl activity, and as such useful in the preparation of medicaments for any condition which may benefit from the inhibition of Sirtl. These not limitedly include cancer, metabolic diseases such as metabolic syndrome, type I diabetes or type II diabetes, obesity, dislipidemia, hyperlipidemia, Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, neurodegenerative conditions that are caused at least in part by polyglutamine aggregation, such as Huntington's disease, spinalbulbar muscular atrophy (SBMA or Kennedy's disease) dentatorubro-pallidoluysian atrophy (DRPLA), spinocerebellar ataxia 1 (SCA1), spinocerebellar ataxia 2 (SCA2), Machado-Joseph disease (MJD;SCA3), spinocerebellar ataxia 6 (SCA6), spinocerebellar ataxia 7 (SCA7), and spinocerebellar ataxia 12 (SCA12).
Compound (I) can be isolated, depending on the method of preparation, in crystalline form A or В or in amorphous form.
Form A is a solvent-free (Fig. 1), non hygroscopic (Fig. 2) form that can be obtained by crystallisation from isopropanol or by concentration at room temperature from various polar protic solvents such as methanol, ethanol, isopropanol or water, as well as from ethyl acetate. Form A is characterised by - an X ray diffraction pattern shown in Fig. 3 having prominent peaks as set out in table 1 below:
Table 1
- An IR absorbtion spectrum shown in Fig. 4 having characteristic peaks expressed in cm'1 at approximately 3448, 3307, 3277, 1649, 1306 and 772 - A Raman spectrum shown in Fig. 5 having characteristic peaks expressed in cm'l at approximately 3450, 3050, 1649, 1616, 1476, 1307, 1194, 901, 831, 323 and 197. (The term approximately means in this context that the values can vary, e.g. by up to +/- 4 cm'1)
- A melting point of about 183°C
Form В (TGA and DSC curves in Fig. 6) is a non hygroscopic form (fig. 7) that can be obtained by evaporation at room temperature from acetone or MEK (methyl ethyl ketone), or a mixture of solvents which contain acetone or МЕК.
Form В is characterised by: - an X ray diffraction pattern shown in Fig. 8 having prominent peaks as set out in table 2 below
Table 2
- An IR absorbtion spectrum shown in Fig. 9 having characteristic peaks expressed in cm'1 at approximately 3389, 1683, 1405 and 1313 - A Raman spectrum shown in Fig. 10 having characteristic peaks expressed in cm'l at approximately 1712, 1623, 1485, 1313, 1163, 843, 339 and 212. (The term approximately means in this context that the values can vary, e.g. by up to +/- 4 cm'1)
- A melting point of about 165°C
The amorphous form of (I) is characterised by the lack of sharp X-ray diffraction peaks in its XRPD pattern (Fig. 11) and can be obtained by cryogrinding.
The amorphous form can easily be converted into form A or into form В. This can be achieved by slurrying in ethanol or acetone, towards forms A or B, respectively. In turn, form В can be easily converted into form A. This can be achieved by slurrying form В in water at various temperatures.
Form A is non hygroscopic (Fig. 2), stable over time (Tables 3 and 4) and it is suitable for use in pharmaceutical compositions.
Form A can be suitably formulated into various pharmaceutically acceptable preparations, which are preferably for oral administration. A three-step method for the preparation of (I) is described in Napper et al. (2005).
This method is however not amenable to large scale production. One drawback of the known method is the need for chromatographic purification of the intermediates. Another drawback is the use of ethyl ether as a solvent in the first step, which is hazardous on technical scale. Another major limiting factor is the presence of a highly exothermic second step, which is incompatible with the safety requirements of large scale production. Particularly, stage 2 of the process disclosed in Napper et al, wherein a compound of formula Г-a wherein R is ethyl is converted into a compound of formula I-b’ using unsolvated reactants, involves an uncontrollable exotherm which makes the process unsafe on a technical scale.
DESCRIPTION OF THE INVENTION
The present invention provides a process for obtaining compound (I) which avoids the drawbacks in the prior art and which is conveniently applicable on a technical scale.
We have surprisingly found that the reaction between compound (Г-а) and 4-chloroaniline can be carried out in far safer conditions under heating using a solvent which forms an azeotropic mixture with water.
Accordingly, in a first embodiment the invention provides a process for the preparation of intermediate (Г-b) according to the following scheme:
wherein R is Ci-C6 linear, branched or cyclic alkyl chain and preferably ethyl, said process being characterised in that the reaction between (Г-а) and 4-chloroaniline is performed under heating in a solvent which forms an azeotropic mixture with water and which is preferably selected from xylenes, chlorobenzene, cyclohexane, ethyl acetate, MTBE, toluene and preferably ethanol.
When the conditions described above are used, the process can be lengthy: we have determined that up to 56 hrs can be necessary for a kg-scale reaction to reach completion. We have found that the elimination of water by azeotropic distillation significantly reduces the reaction time (see examples 3 and 4). Best results can be achieved by performing at least two azeotropic distillations of the reaction mixture.
Accordingly, in a preferred embodiment, the process for the preparation of compound (Г-b) as above defined further comprises removing water from the reaction mixture by azeotropic distillation.
We have also determined that the chromatographic purification of (I’-b) can be avoided if the crude product is crystallised from an apolar solvent such as cyclohexane. Accordingly, in a further preferred embodiment, the crude mixture containing compound (Г-b) obtained by the invention process is crystallized from an apolar solvent, preferably from cyclohexane (see examples 3 and 4).
In a further embodiment of the invention, the intermediate (Г-b) is converted into compound (I):
by reaction with ammonia in a suitable solvent. The skilled chemist may easily replace this step by a two-step procedure involving the reaction between (Г-b) and suitably protected amine derivative followed by deprotection of this amine in order to obtain (I).
The choice of the solvent in which to perform conversion step (I’-b)—>(I) influences the reaction yield. We have determined that this step should not be performed using water as solvent because part of the ester (Г-b) converts to its free acid equivalent. Particularly suitable solvents in which to perform the reaction are ethanol and methanol, the latter being preferred in that, the reaction reaches completion in shorter times. When using methanol, ammonia may be added either pure or as a methanolic solution, the former being preferred (examples 5 and 6).
Compound (I) may be further processed by: a) removing any unreacted ammonia from the reaction mixture b) precipitating the compound from the reaction mixture c) recrystallising the precipitate from a suitable solvent so as to obtain the compound of formula (I) in the crystalline form A.
Step a) may be performed by evaporation under reduced pressure and/or heating and/or sparging the reaction mixture with an inert gas.
Step b) may be performed by addition of water to the reaction mixture.
Exemplary suitable solvents in step c) are ethyl acetate, water, methanol, ethanol and isopropanol, with isopropanol being preferred.
Should one wish to obtain GMP-grade form A, it may be preferable to perform two or more subsequent crystallizations. We have determined that GMP-grade material can be obtained after only two re-crystallisations if the crude precipitate ensuing from step b) is crystallised from MEK/cyclohexane before performing step c) (example 7).
Thus, the process of the invention does not require chromatographic purification of the final product and allows to obtain (I) in crystalline form A.
The starting compound (Г-a) can be obtained by reacting equimolar amounts of bromine and a compound of formula Г-c, at 0-5°C in DCM:
Alternatively, the compound (I-a’) is obtained by slow addition, under stirring, of gaseous bromine to a solvent-free equimolar amount of compound (Г-с), atO - 10°C.
In either way, the use of the solvent ethyl ether, which is hazardous on a technical scale, is avoided, and the impact on the environment is reduced by eliminating the amount of chlorinated solvent waste.
The invention will be further illustrated by the following examples and attached figures.
DESCRIPTION OF THE FIGURES
Fig. 1: DSC (upper) and TGA (lower) curves of form A Fig. 2: Absorption-Desorption spectrum of form A Fig. 3: XPRD spectrum of form A Fig. 4: IR spectrum of form A Fig. 5: Raman spectrum of form A
Fig.6: DSC (upper) and TGA (lower) curves of form В isolated from acetone.
Fig. 7: Absorption-desorption spectrum of form В
Fig. 8: XPRD spectmm of Form В
Fig. 9: IR spectrum of form В
Fig. 10: Raman spectmm of form В
Fig. 11: XPRD spectmm of the amorphous form
EXAMPLES
Example 1: stage 1 procedure ((I’-c)—»(I’-a)) involving DCM as solvent
Ethyl-2-cyclohexanone-carboxylate (2 kg, 11.75 mol) and DCM (6L) were charged to a 20L jacketed reactor and cooled to 0-5°C. Bromine (1877.8 g, 11.75 mol, 1 equiv) was pumped in over 2.5 hours maintaining the reaction temperature at 0-5°C. The reaction solution was then held at 0-5°C for a further 2 hours before sampling. (GC area%:1.7% ethyl-2-cyclohexanone-carboxylate, 76.4% 3-bromo-2-oxo-cyclohexanecarboxylic acid ethyl ester).
The reaction was quenched with water (5L) allowing the exotherm to take the temperature to 20-25°C. The organic layer (bottom, hazy yellow, 10.78 kg) was separated form the clear colourless aqueous layer (5.97 kg).
The organic layer was washed with sat. sodium bicarbonate solution (4 kg) and then separated (organic (10.43 kg) and aqueous (4.2 kg)). The organic layer was stripped to an oil under reduced pressure and ethanol (2L) charged. The ethanol was then removed by distillation at reduced pressure to give a orange oil (2950.9 g).
Yield of crude product = 100.8%
Purity (GC area%) = 2.4% keto ester, 6.6% unkn (9.6min), 89.6% 3-bromo-2-oxo-cyclohexanecarboxylic acid ethyl ester.
Example 2: stage 1 procedure with no solvent
Ethyl-2-cyclohexanone-carboxylate (497 kg) is cooled down to 0°C and bromine is added over 9h while stirring, keeping the temperature at 0°±10°C. After complete addition, the mixture is stirred at 0°C for 4 h (content of starting material 2.8% by GC), then it is warmed up to 20°C and nitrogen is bubbled through the reaction mixture to drive off the HBr gas. Then the reaction mixture is stirred for another 25 h at 5°C (content of HBr 0.8% by titration). 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-l-carboxylic acid ethyl ester is drummed off and stored at 0°C prior to use as such in the subsequent step.
Yield of crude product: 730.4 kg (100%)
Purity (determined by 'H-NMR with 3,4-Dimethoxybenzene as internal standard). 87% w/w.
Example 3: stage 2 procedure ((I’-a)—»(I’-b)) with no intermediate azeotropic distillation 4-chloroaniline (2473.5 g, 19.39 mol, 2.1 equiv) and ethanol (13.8L) were charged to a 20L jacketed reactor and heated to reflux (80°C). The 3-bromo-2-oxo-cyclohexanecarboxylic acid ethyl ester (2300 g, 9.23 mol, 1.0 equiv) was added over ~3hrs maintaining reflux (a sample was taken: 338.3 g, 2.2% of yield). The reaction was held at reflux and was deemed complete after 56 hours (HPLC area%: 2.9% intermediates, 88.7% 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-l-carboxylic acid ethyl ester).
The solvent was removed by distillation keeping the temperature below 50°C. Cyclohexane (10.58L) and water (4.6L) was added to the residue and the mixture heated to 50-55°C. The dark purple aqueous layer was separated from the dark brown organic layer. The organic layer was washed with 2M HC1 (3x4.6L) at 50-55°C and the organic layer sampled to determine aniline content (non-detected by LCMS, spec <1% wrt 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-1-carboxylic acid ethyl ester). The organic layer was washed with sat. sodium chloride solution (lx4.6L) followed by sat. sodium bicarbonate solution (1x4.6L) and finally water (1x4.6L) at 50-55°C. The dark brown solution was then dried by the azeotropic removal of water (KF =0.02% spec <0.5%). The solution was then cooled to 15°C and then isolated by filtration. The filter cake was washed with cold cyclohexane (15°C) and the damp green product was dried at 50°C for 16hours. The 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-1-carboxylic acid ethyl ester solid was isolated as a green crystalline solid (1636 g).
Wt = 1636 g, Yield = 63.8% Purity LCMS (210 nm) = 99.4%, HPLC (215 nm) = 97.6%.
Example 4: stage 2 procedure with two intermediate azeotropic distillations A solution of 4-chloroaniline (266 kg) in EtOH (1425 1) was warmed to reflux (76°C) and 3-bromo-2-oxo-cyclohexanecarboxylic acid ethyl ester (240 kg) was added over lh 40 min, while stirring at reflux (76 to 80°C). Reflux (80°C) was maintained for 5h, then EtOH (200 1) was added, and solvent (200 1) was distilled off. Then EtOH (140 1) was added followed by distilling off solvent (140 1). The reaction mixture was then heated at reflux for another 17 h, after which the solvent (1400 1) is distilled off at reduced pressure and 60±5°C.
Then cyclohexane (1122 1) is added while stirring at 45 - 59°C. To the cyclohexane solution, stirred at 52 - 59°C is then added water (482 1). The phase are separated,and the aqueous phase is discarded. The organic phase (kept at 54 - 58°C) is then washed five times with a mixture of water (184 1) and cone. HC1 (48 1), followed by washing with water (240 1), sodium bicarbonate (25 kg) in water (250 1) solution and finally water (480 1).
Then more cyclohexane (453 1) is added to the warm (55 - 57°C) solution, followed by evaporation of solvent (453 1) to azeotropically remove water. Then the reaction mixture is stepwise cooled to 40°C over 2h, to 10°C over 2h and kept at 5 - 10°C for 1.5h to crystallise 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-1-carboxylic acid ethyl ester. After centrifugation, the solid is recrystallised once more from cyclohexane (731 1), centrifuged and drummed off without further drying for use in the subsequent step. Yield: 183 kg. LOD (determined by lab sample): 10%, Yield = 70.8%, Purity (HPLC): 99.4%.
Example 5: stage 3 procedure ((I’-b)^(I)) using ammonia in methanolic solution 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-l-carboxylic acid ethyl ester and ammonia in methanol (7N) were charged to the 3 litre autoclave and heated to 60-65°C (a pressure of 2.3 bar was reached). The reaction mixture was then held at this temperature for 48 hours then cooled to 20-25°C and sampled for completion. The reaction was deemed complete (LCMS(210 nm,area%): 6-chloro-2,3,4,9-tetrahydro-1 H-carbazole-1 -carboxylic acid methyl ester 1.4%, 6-chloro-2,3,4,9-tetrahydro-l H-carbazole-1-carboxylic acid amide 96.7%: HPLC(215nm,area%): 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-1-carboxylic acid methyl ester 0.6%, 6-chloro-2,3,4,9-tetrahydro- Ш-carbazole-l-carboxylic acid ethyl ester 0.3%, 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-1-carboxylic acid amide 97.8%). The reaction solution was transferred to a 3L flange reactor and nitrogen was bubbled through the solution via a sparge tube for 3 hours. During this process the product precipitated out of solution. The mixture was then heated to reflux and methanol (505 ml) was removed by distillation at atmospheric pressure (distillation started at 42°C head T/46°C pot T and was terminated at 60°C head T/63°C pot T). Water (270ml) was then added slowly over 15 minutes maintaining the reaction temperature at reflux (the product precipitated during the addition). The mixture was cooled to 0-5°C and held at this temperature for 1 hour. The product was isolated by filtration and the damp filter cake washed with cold (5°C) methanol/ water 1:1 (120 ml). The crude 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-l-carboxylic acid amide was isolated as a damp grey crystalline solid (305.59 g).
Wt = 305.59 g, Str = 75.6% (taking LOD into account), Yield = 231.02 g (86%).
The damp crude 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-l-carboxylic acid amide (305.59 g, 231.02 g 100%) was heated to reflux (82°C, product dissolves at ~74°C) in 2-propanol (1975.2 g). 2-Propanol (200 ml) was removed by distillation at atmospheric pressure and the solution cooled to ~60°C. The solution was then seeded and the product crystallised. The mixture was then held at 60°C for 1 hour and then cooled slowly to 0-5°C. The mixture was held at this temperature for 1 hour. The product was isolated by filtration and the damp filter cake washed with cold (5°C) IPA (250 ml). The damp solid was then dried for 16 hours at 60°C to give an off-white crystalline solid (189.95 g).
Wt = 189.95 g (82.2% for crystallisation, 70.7% for 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-1-carboxylic acid amide overall).
Purity LCMS (21 Опт) = 100% HPLC (215nm) = 100%
Moisture content (by KF) = 0.05% LOD = 0.5%
Example 6: stage 3 procedure using pure ammonia A solution of 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-l-carboxylic acid ethyl ester (183 kg) in MeOH (850 1) in a pressure reactor was cooled to 5±5°C, and ammonia (328 kg) was added over 4 h while stirring at to 5±5°C. Then the temperature was increased to 45±5°C (4 bar internal pressure) and kept at this temperature for 47 h (HPLC check: no s.m. left).
The solution was cooled to 0 - 10°C, filtered into a non-pressure reactor and warmed to 45±5°C over 4 h and kept at this temperature for another 12 h to gas off the excess ammonia. After charcoal treatment (8 kg) at 60°C, filtration, washing with MeOH (150 1) and evaporation of solvent (300 1), water (200 1) is added and the mixture is cooled to 3±3°C to precipitate 6-chloro-2,3,4,9-tetrahydro-lH-carbazole-l-carboxylic acid amide product. The solid is centrifuged and drummed off for the subsequent recrystallisation. Yield: 142 kg. With a LOD = 16% this corresponds to 119 kg dry material (81.3% of theory).
This material, dissolved in in methyethyl ketone (407 kg) is heated to reflux, and cyclohexane is added at to 68 -77°C over 15 min. The mixture is stirred at to 68°C for 1.5h, then it is cooled to 0°C over 2h. The solid is isolated by centrifugation (146 kg, wet).
The solid is suspended in 2-propanol heated to reflux within 2h 20 min (complete dissolution), then cooled to 0±5°C over 2.5h and kept at to 0±5°C for lh. The solid is centrifuged and then dried for 18 in a paddle dryer (to 20 - 49°C, 28 - 86 mbar) to give the final product (88.3 kg).
Example 7: crystallization from Cyclohexane/MEK followed by re-crystallisatrion from isopropanol affords higher purity material than 2 subsequent crystallisations from isopropanol
Aliquots of the same batch ensuing from step b) (claim 8) were crystallised twice as set out the table below
Tables 3 and 4: Form A stability data: Compound (I) in the form of Form A was packed in double bagged polythene bags (fastened with cable/security ties) and placed inside 3.6L Curtec kegs (HDPE) to simulate typical drug substance storage conditions. Stability tests were performed at 40°C/75% RH (table 3) and 25°C/60% RH (table 4).
Table 3
Table 4

Claims (14)

1. A process for the preparation of compound (Г-b) according to the following scheme:
wherein R is Ci-C6 linear, branched or cyclic alkyl chain and preferably ethyl, said process being characterised in that the reaction between (Г-а) and 4-chloroaniline is performed under heating in a solvent which forms an azeotropic mixture with water.
2. A process according to claim 1, wherein said solvent is selected from xylenes, chlorobenzene, cyclohexane, ethyl acetate, MTBE, toluene and preferably ethanol.
3. A process according to claim 1, wherein the water ensuing from said reaction is removed by azeotropic distillation.
4. A process according to claims 1-3, further comprising crystallization of compound (Г-b) from an apolar solvent.
5. A process according to claim 4, wherein said apolar solvent is cyclohexane.
6. A process for the preparation of compound (I)
comprising the conversion of compound (Г-а) into compound (Г-b) as defined in claims 1-5, followed by reaction of (Г-b) with ammonia.
7. A process according to claim 6, wherein said reaction is carried out in a solvent selected from ethanol and methanol.
8. A process according to claims 6-7, wherein compound (I) is further processed by: a) removing any unreacted ammonia from the reaction mixture; b) precipitating compound (I) from the reaction mixture; c) recrystallising the precipitate from a suitable solvent so as to obtain compound (I) in crystalline form A.
9. A process according to claim 8, wherein step a) is performed by evaporation under reduced pressure and/or heating and/or sparging the reaction mixture with an inert gas.
10. A process according to claim 8, wherein step b) is performed by addition of water to the reaction mixture.
11. A process according to claim 8, wherein the solvent used in step c) is selected from ethyl acetate, water, methanol, ethanol and preferably isopropanol.
12. A process according to claim 8 and 11, further comprising crystallizing the crude precipitate ensuing from step b) from a MEK/cyclohexane solvent mixture before performing step c).
13. A process according to claims 1 or 6, wherein compound (Г-а) is obtained by reacting equimolar amounts of bromine and a compound of formula (Г-с), at 0-5°C in DCM:
14. A process according to claims 1 or 6, wherein compound (I-a’) is obtained by slow addition, under stirring, of gaseous bromine to a solvent-free equimolar amount of compound (Г-с), at 0 - 10°C.
AU2012324824A 2011-10-20 2012-10-19 Processes for the preparation of 6-Chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and of its precursors Active AU2012324824B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP11185959 2011-10-20
EP11185959.1 2011-10-20
PCT/EP2012/070769 WO2013057258A1 (en) 2011-10-20 2012-10-19 Processes for the preparation of 6-chloro-2,3,4,9-tetrahydro-1h-carbazole-1-carboxamide and of its precursors

Publications (2)

Publication Number Publication Date
AU2012324824A1 AU2012324824A1 (en) 2014-05-08
AU2012324824B2 true AU2012324824B2 (en) 2016-08-11

Family

ID=47143859

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2012324824A Active AU2012324824B2 (en) 2011-10-20 2012-10-19 Processes for the preparation of 6-Chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and of its precursors

Country Status (17)

Country Link
US (2) US20140303382A1 (en)
EP (1) EP2768807B1 (en)
JP (1) JP6142459B2 (en)
CN (1) CN103958467B (en)
AR (1) AR088377A1 (en)
AU (1) AU2012324824B2 (en)
BR (1) BR112014009297A2 (en)
CA (1) CA2852797C (en)
ES (1) ES2585077T3 (en)
HK (1) HK1200452A1 (en)
HR (1) HRP20160918T1 (en)
HU (1) HUE029282T2 (en)
IL (1) IL232058A (en)
PL (1) PL2768807T3 (en)
PT (1) PT2768807T (en)
SI (1) SI2768807T1 (en)
WO (1) WO2013057258A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201312768D0 (en) * 2013-07-17 2013-08-28 Ge Healthcare Ltd Work-up procedure

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008019825A1 (en) * 2006-08-14 2008-02-21 Santhera Pharmaceuticals (Schweiz) Ag Use of tricyclic indole derivatives for the treatment of muscular diseases

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3769298A (en) * 1971-04-02 1973-10-30 Pfizer Substituted aminomethylcarbazoles
DE2337154C2 (en) * 1973-07-18 1986-04-24 Schering AG, 1000 Berlin und 4709 Bergkamen New carbazole derivatives
US4057640A (en) * 1973-07-18 1977-11-08 Schering Aktiengesellschaft 5,6,7,8:Tetrahydrocarbazole-1-carboxylic acid derivatives
DE2431292A1 (en) * 1974-06-27 1976-01-15 Schering Ag Anti-inflammatory carbazole derivs - specif. e.g. 1-ethoxycarbonyl-6-methoxy-carbazole and 8-methyl-carbazole-1-carboxylic acid
BG25793A3 (en) 1973-07-18 1978-12-12 Schering Ag A method of obtaining carbasole derivatives
US3907054A (en) 1974-06-13 1975-09-23 Caterpillar Tractor Co Diagonal brace bearing for crawler-type tractors
GB1504707A (en) 1974-08-07 1978-03-22 Bellon Labor Sa Roger 1-aryl-2-oxo-2,4,5,6,7,7a-hexahydroindoles
DZ2769A1 (en) 1998-04-17 2003-12-01 Lilly Co Eli Substituted tricyclics.
DZ2770A1 (en) 1998-04-17 2003-12-01 Lilly Co Eli Process for the preparation of 1h-indol-3 glyoxamides substituted in position 4.
AR038136A1 (en) * 2002-01-24 2004-12-29 Merck Frosst Canada Inc CYCLALCANINDOLS WITH REPLACEMENT WITH FLUOR COMPOSITIONS CONTAINING THESE COMPOUNDS AND TREATMENT METHODS
CA2509202A1 (en) 2003-01-06 2004-07-29 Eli Lilly And Company Fused heterocyclic derivatives as ppar modulators
FR2849811B1 (en) 2003-01-10 2006-01-21 Renault Sas LONGITUDINAL CONTROL SYSTEM FOR MAINTAINING A VEHICLE WITH AN AUTOMATIC GEARBOX
JP4908215B2 (en) * 2003-09-12 2012-04-04 エリクサー ファーマスーティカルズ インコーポレイテッド Treatment of disease
US20060074124A1 (en) * 2003-09-12 2006-04-06 Andrew Napper Methods of treating a disorder
US7534264B2 (en) * 2004-01-28 2009-05-19 Ultradent Products, Inc. Delivery system for bone growth promoting material
CA2553814A1 (en) * 2004-01-29 2005-08-11 Elixir Pharmaceuticals, Inc. Treating a viral disorder
US7104702B2 (en) 2004-03-24 2006-09-12 Corning Cable Systems Llc Field installable optical fiber connector
US20060003065A1 (en) 2004-07-01 2006-01-05 Kateman Paul R Dry-base aerated food product dispensing method and apparatus
CA2588384A1 (en) * 2004-11-23 2006-06-22 Ptc Therapeutics, Inc. Tetrahydrocarbazoles as active agents for inhibiting vegf production by translational control
AR052674A1 (en) * 2005-02-17 2007-03-28 Wyeth Corp DERIVATIVES OF INDOL, BENZOTIOFEN, BENZOFURAN AND INDENO CICLOALQUILCONDENSADOS
EP1865913A1 (en) * 2005-03-11 2007-12-19 Elixir Pharmaceuticals, Inc. Sirt inhibitors that bind to nad
US20090022694A1 (en) * 2005-10-18 2009-01-22 Distefano Peter Sirt1 inhibition
DE102006029320B3 (en) * 2006-06-23 2007-10-11 Röhm Gmbh Continuous preparation of unsaturated carboxylic acid anhydride comprises anhydrating aliphatic carboxylic acid anhydride with carboxylic acid in rectification column, recycling the non-reacted educt in reaction medium and collecting

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008019825A1 (en) * 2006-08-14 2008-02-21 Santhera Pharmaceuticals (Schweiz) Ag Use of tricyclic indole derivatives for the treatment of muscular diseases

Also Published As

Publication number Publication date
BR112014009297A2 (en) 2017-04-11
PT2768807T (en) 2016-08-08
CN103958467B (en) 2016-10-12
AU2012324824A1 (en) 2014-05-08
US20140303382A1 (en) 2014-10-09
HUE029282T2 (en) 2017-02-28
CA2852797C (en) 2020-02-04
JP2014530839A (en) 2014-11-20
ES2585077T3 (en) 2016-10-03
EP2768807A1 (en) 2014-08-27
AR088377A1 (en) 2014-05-28
HRP20160918T1 (en) 2016-09-23
CA2852797A1 (en) 2013-04-25
JP6142459B2 (en) 2017-06-07
US20160304455A1 (en) 2016-10-20
WO2013057258A1 (en) 2013-04-25
IL232058A0 (en) 2014-05-28
PL2768807T3 (en) 2017-03-31
US10329254B2 (en) 2019-06-25
CN103958467A (en) 2014-07-30
HK1200452A1 (en) 2015-08-07
SI2768807T1 (en) 2016-09-30
EP2768807B1 (en) 2016-05-04
IL232058A (en) 2016-02-29

Similar Documents

Publication Publication Date Title
US9102658B2 (en) Process for the preparation of (5-fluoro-2-methyl-3-quinolin-2-ylmethyl-indol-1-y1)-acetic acid esters
WO2006061364A1 (en) Process for the preparation of carvedilol and its enantiomers
WO2018141642A1 (en) Process for the preparation of 2-chloro-4-fluoro-5-nitrobenzotrichloride
AU2012324824B2 (en) Processes for the preparation of 6-Chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and of its precursors
US7875730B2 (en) Process for manufacture of racemic Carvedilol
ES2955674T3 (en) Method for producing cis-alkoxy-substituted spirocyclic derivatives 1-h-pyrrolidine-2,4-dione
US20120253051A1 (en) Process for the preparation of ropinirole and salts thereof
US20190314385A1 (en) Process for Preparation of Chlorpromazine or its Pharmaceutically Acceptable Salts
EP2804607B1 (en) Synthesis of raltegravir
WO2007042848A2 (en) Process for the preparation of carbidopa
US20110046416A1 (en) Process for preparation of benzphetamine and its pharmaceutically acceptable salts
US9499491B2 (en) One pot process for the preparation of telmisartan
TW201323407A (en) Process for the preparation of 6-Chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide and intermediates thereof
CN107207435B (en) Process for preparing 4-cyanopiperidine hydrochloride
WO2019167058A1 (en) An improved process for the preparation of propiomazine maleate
JP5613509B2 (en) Method for obtaining fexofenadine monohydrochloride
WO2011124638A1 (en) Pimobendan manufacturing process
JP2012087100A (en) Method for producing fexofenadine monohydrochloride of form i
CZ20023312A3 (en) Preparation and isolation of 2-substituted-3-pyridylcarboxylic acids, their carboxylic salts and reduction products
CZ296056B6 (en) Process for preparing oxybutynin
WO2016135616A1 (en) An improved process for the preparation of bisoprolol and its intermediate
WO2014184754A1 (en) Method for preparing anastrozole for pharmaceutical purposes
JPH0585985A (en) Production of optically active atrolactinic acid and intermediate for production

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
PC Assignment registered

Owner name: AOP ORPHAN PHARMACEUTICALS

Free format text: FORMER OWNER(S): SIENA BIOTECH S.P.A.

PC Assignment registered

Owner name: IPSOL AG

Free format text: FORMER OWNER(S): AOP ORPHAN PHARMACEUTICALS